Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutrients ; 15(18)2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37764787

RESUMO

We investigated the effects of two dosing regimens of two multi-strain probiotic products on the gut microbiota of breastfed infants, including the transfer of the dosed strains and clinical outcomes. In forty-seven dyads, infants were either exposed through maternal intake (MS) of Lactobacillus acidophilus LA-5, Bifidobacterium animalis subsp. lactis BB-12, Lacticaseibacillus rhamnosus LGG, and Bifidobacterium longum subsp. infantis Bifin02 from gestational week thirty-three until four weeks after birth (n = 24) or dosed directly (IS) with the same strains except for LA-5 starting within 24 h after birth until day 28 (n = 23). Infant stool samples were collected on day 0, 14, 28, and 42 after birth. Gastrointestinal symptoms were assessed by parents using an electronic diary. Microbiota composition was determined using 16S rRNA sequencing, and strain recovery was analyzed by qPCR. Notably, 100% of the IS infants were colonized with Bifin02 after 14 days as opposed to only 25% of the MS infants. Mean stool frequency was significantly lower in IS infants compared to MS infants and IS infants had softer stools on day 14, 28, and 42. A significantly steeper slope of progression of inconsolable crying and fussing was observed in MS infants compared to IS infants. In conclusion, direct infant seeding induced a faster increase in fecal bifidobacteria abundancy and Bifin02 recovery compared to dosed through the maternal intake.


Assuntos
Bifidobacterium animalis , Microbiota , Probióticos , Feminino , Humanos , Lactente , Aleitamento Materno , RNA Ribossômico 16S/genética , Lactobacillus acidophilus , Fezes/microbiologia , Bifidobacterium longum subspecies infantis/genética
2.
Nat Commun ; 14(1): 1349, 2023 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-36906612

RESUMO

Preterm infants with very low birthweight are at serious risk for necrotizing enterocolitis. To functionally analyse the principles of three successful preventive NEC regimens, we characterize fecal samples of 55 infants (<1500 g, n = 383, female = 22) longitudinally (two weeks) with respect to gut microbiome profiles (bacteria, archaea, fungi, viruses; targeted 16S rRNA gene sequencing and shotgun metagenomics), microbial function, virulence factors, antibiotic resistances and metabolic profiles, including human milk oligosaccharides (HMOs) and short-chain fatty acids (German Registry of Clinical Trials, No.: DRKS00009290). Regimens including probiotic Bifidobacterium longum subsp. infantis NCDO 2203 supplementation affect microbiome development globally, pointing toward the genomic potential to convert HMOs. Engraftment of NCDO 2203 is associated with a substantial reduction of microbiome-associated antibiotic resistance as compared to regimens using probiotic Lactobacillus rhamnosus LCR 35 or no supplementation. Crucially, the beneficial effects of Bifidobacterium longum subsp. infantis NCDO 2203 supplementation depends on simultaneous feeding with HMOs. We demonstrate that preventive regimens have the highest impact on development and maturation of the gastrointestinal microbiome, enabling the establishment of a resilient microbial ecosystem that reduces pathogenic threats in at-risk preterm infants.


Assuntos
Microbioma Gastrointestinal , Recém-Nascido Prematuro , Lactente , Recém-Nascido , Humanos , Feminino , RNA Ribossômico 16S/genética , Ecossistema , Intestinos , Fezes/microbiologia , Bifidobacterium longum subspecies infantis/genética
3.
mSystems ; 7(5): e0034322, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36094076

RESUMO

Bifidobacterium longum subsp. infantis is a prevalent beneficial bacterium that colonizes the human neonatal gut and is uniquely adapted to efficiently use human milk oligosaccharides (HMOs) as a carbon and energy source. Multiple studies have focused on characterizing the elements of HMO utilization machinery in B. longum subsp. infantis; however, the regulatory mechanisms governing the expression of these catabolic pathways remain poorly understood. A bioinformatic regulon reconstruction approach used in this study implicated NagR, a transcription factor from the ROK family, as a negative global regulator of gene clusters encoding lacto-N-biose/galacto-N-biose (LNB/GNB), lacto-N-tetraose (LNT), and lacto-N-neotetraose (LNnT) utilization pathways in B. longum subsp. infantis. This conjecture was corroborated by transcriptome profiling upon nagR genetic inactivation and experimental assessment of binding of recombinant NagR to predicted DNA operators. The latter approach also implicated N-acetylglucosamine (GlcNAc), a universal intermediate of LNT and LNnT catabolism, and its phosphorylated derivatives as plausible NagR transcriptional effectors. Reconstruction of NagR regulons in various Bifidobacterium lineages revealed multiple potential regulon expansion events, suggesting evolution from a local regulator of GlcNAc catabolism in ancestral bifidobacteria to a global regulator controlling the utilization of mixtures of GlcNAc-containing host glycans in B. longum subsp. infantis and Bifidobacterium bifidum. IMPORTANCE The predominance of bifidobacteria in the gut of breastfed infants is attributed to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). Thus, individual HMOs such as lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT) are considered promising prebiotics that would stimulate the growth of bifidobacteria and confer multiple health benefits to preterm and malnourished children suffering from impaired (stunted) gut microbiota development. However, the rational selection of HMO-based prebiotics is hampered by the incomplete knowledge of regulatory mechanisms governing HMO utilization in target bifidobacteria. This study describes NagR-mediated transcriptional regulation of LNT and LNnT utilization in Bifidobacterium longum subsp. infantis. The elucidated regulatory network appears optimally adapted to simultaneous utilization of multiple HMOs, providing a rationale to add HMO mixtures (rather than individual components) to infant formulas. The study also provides insights into the evolutionary trajectories of complex regulatory networks controlling carbohydrate metabolism in bifidobacteria.


Assuntos
Bifidobacterium , Leite Humano , Lactente , Recém-Nascido , Feminino , Criança , Humanos , Bifidobacterium/genética , Leite Humano/química , Prebióticos/análise , Oligossacarídeos/análise , Polissacarídeos/análise , Bifidobacterium longum subspecies infantis/genética
4.
Front Immunol ; 13: 871705, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35860248

RESUMO

Aim: Our objective was to investigate whether Bifidobacterium infantis inhibits PI3K-Akt-mTOR signaling and upregulates Foxp3 expression through PD-L1 and to explore the possible mechanism of action of B. infantis in cellular immunosuppression. Method: The effects of B. infantis supernatant on PD-L1, PD-1, Foxp3, and the PI3K-Akt-mTOR signaling pathway were observed by culturing HCT-116 cells. Simultaneously, the effects of blocking PD-L1 on PD-1, on Foxp3 protein and mRNA, and on the PI3K-Akt-mTOR signaling pathway protein were observed. Results: B. infantis supernatant was able to upregulate the protein and mRNA expression of PD-L1 and Foxp3 and downregulate the phosphorylated protein expression of PI3K, Akt, and mTOR (P < 0.05); however, for PI3K, Akt, and mTOR, there was no change in the total protein expression. After the blocking of PD-L1, the stimulatory effect of B. infantis supernatant on Foxp3 and the inhibitory effect on the phosphorylated protein expression of PI3K, Akt, and mTOR were weakened. Conclusion: B. infantis may inhibit the PI3K-Akt-mTOR signaling pathway and promote the expression of Foxp3 through PD-L1, which may be a target via which B. infantis exerts its immunosuppressive effect.


Assuntos
Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/metabolismo , Colo/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Morte Celular Programada 1 , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
5.
Science ; 376(6598): 1220-1223, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35679413

RESUMO

Infant microbiome assembly has been intensely studied in infants from industrialized nations, but little is known about this process in nonindustrialized populations. We deeply sequenced infant stool samples from the Hadza hunter-gatherers of Tanzania and analyzed them in a global meta-analysis. Infant microbiomes develop along lifestyle-associated trajectories, with more than 20% of genomes detected in the Hadza infant gut representing novel species. Industrialized infants-even those who are breastfed-have microbiomes characterized by a paucity of Bifidobacterium infantis and gene cassettes involved in human milk utilization. Strains within lifestyle-associated taxonomic groups are shared between mother-infant dyads, consistent with early life inheritance of lifestyle-shaped microbiomes. The population-specific differences in infant microbiome composition and function underscore the importance of studying microbiomes from people outside of wealthy, industrialized nations.


Assuntos
Bifidobacterium longum subspecies infantis , Países em Desenvolvimento , Microbioma Gastrointestinal , Estilo de Vida , Bifidobacterium longum subspecies infantis/classificação , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/isolamento & purificação , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/genética , Genoma Bacteriano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Leite Humano/microbiologia , Tanzânia
6.
Gut Microbes ; 13(1): 1973835, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34553672

RESUMO

Certain existing prebiotics meant to facilitate the growth of beneficial bacteria in the intestine also promote the growth of other prominent bacteria. Therefore, the growth-promoting effects of ß-galactosides on intestinal bacteria were analyzed. Galactosyl-ß1,4-l-rhamnose (Gal-ß1,4-Rha) selectively promoted the growth of Bifidobacterium. Bifidobacterium longum subsp. longum 105-A (JCM 31944) has multiple solute-binding proteins belonging to ATP-binding cassette transporters for sugars. Each strain in the library of 11 B. longum subsp. longum mutants, in which each gene of the solute-binding protein was disrupted, was cultured in a medium containing Gal-ß1,4-Rha as the sole carbon source, and only the BL105A_0502 gene-disruption mutant showed delayed and reduced growth compared to the wild-type strain. BL105A_0502 homolog is highly conserved in bifidobacteria. In a Gal-ß1,4-Rha-containing medium, Bifidobacterium longum subsp. infantis JCM 1222T, which possesses BLIJ_2090, a homologous protein to BL105A_0502, suppressed the growth of enteric pathogen Clostridioides difficile, whereas the BLIJ_2090 gene-disrupted mutant did not. In vivo, administration of B. infantis and Gal-ß1,4-Rha alleviated C. difficile infection-related weight loss in mice. We have successfully screened Gal-ß1,4-Rha as a next-generation prebiotic candidate that specifically promotes the growth of beneficial bacteria without promoting the growth of prominent bacteria and pathogens.


Assuntos
Bifidobacterium longum subspecies infantis/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Clostridioides difficile/crescimento & desenvolvimento , Dissacarídeos/farmacologia , Prebióticos/análise , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Bifidobacterium/genética , Bifidobacterium longum subspecies infantis/genética , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Anaerobe ; 68: 102320, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33460787

RESUMO

Bifidobacterium longum subsp. infantis ATCC 15697 has emerged as a model for infant gut-associated bifidobacterial strains. Here we present a genetic system for B. longum subsp. infantis ATCC 15697 using its own DNA restriction-modification systems and create a fucose permease deletion mutant lacking the ability to use free fucose as a carbon source.


Assuntos
Proteínas de Bactérias/genética , Bifidobacterium longum subspecies infantis/enzimologia , Fucose/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Bactérias/metabolismo , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/metabolismo , Deleção de Genes , Proteínas de Membrana Transportadoras/metabolismo
8.
Food Chem ; 346: 128887, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33385916

RESUMO

As interest in probiotics increases, the need for accurate description of probiotic compositions present in commercial products is also increasing. Since Bifidobacterium longum used as probiotics is labeled at species or subspecies levels, a detection method for distinguishing B. longum subsp. longum, infantis, and suis is needed. Thus, we designed three LAMP primer sets for B. longum subspecies. Each primer set was specific for the target subspecies. The detection level was 0.2 pg for each target DNA (about 102 CFU/mL). To apply these LAMP assays to on-site detection, a direct DNA extraction method was optimized and combined with LAMP assay. Finally, direct LAMP assays were used to monitor the presence of B. longum subspecies in 16 probiotic products. They could specifically and sensitively detect target subspecies within approximately 45 min. These rapid on-site detection methods are useful for identifying B. longum subspecies in probiotic products.


Assuntos
Bifidobacterium longum subspecies infantis/isolamento & purificação , Bifidobacterium/isolamento & purificação , DNA Bacteriano/análise , Probióticos/análise , Bifidobacterium/genética , Bifidobacterium longum subspecies infantis/genética , DNA Bacteriano/metabolismo , Humanos , Limite de Detecção , Técnicas de Diagnóstico Molecular , Técnicas de Amplificação de Ácido Nucleico
9.
Benef Microbes ; 12(1): 69-83, 2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33191780

RESUMO

The establishment of the gut microbiota immediately after birth is a dynamic process that may impact lifelong health. At this important developmental stage in early life, human milk oligosaccharides (HMOs) serve as specific substrates to shape the gut microbiota of the nursling. The well-orchestrated transition is important as an aberrant microbial composition and bacterial-derived metabolites are associated with colicky symptoms and atopic diseases in infants. Here, we study the trophic interactions between an HMO-degrader, Bifidobacterium infantis and the butyrogenic Anaerostipes caccae using carbohydrate substrates that are relevant in the early life period including lactose and total human milk carbohydrates. Mono- and co-cultures of these bacterial species were grown at pH 6.5 in anaerobic bioreactors supplemented with lactose or total human milk carbohydrates. A. caccae was not able to grow on these substrates except when grown in co-culture with B. infantis, leading to growth and concomitant butyrate production. Two levels of cross-feeding were observed, in which A. caccae utilised the liberated monosaccharides as well as lactate and acetate produced by B. infantis. This microbial cross-feeding points towards the key ecological role of bifidobacteria in providing substrates for other important species that will colonise the infant gut. The progressive shift of the gut microbiota composition that contributes to the gradual production of butyrate could be important for host-microbial crosstalk and gut maturation.


Assuntos
Bifidobacterium longum subspecies infantis/metabolismo , Clostridiales/metabolismo , Lactose/metabolismo , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/crescimento & desenvolvimento , Reatores Biológicos/microbiologia , Clostridiales/genética , Clostridiales/crescimento & desenvolvimento , Técnicas de Cocultura , Meios de Cultura/metabolismo , Humanos
10.
Nutrients ; 12(11)2020 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-33114073

RESUMO

Dysbiosis is associated with acute and long-term consequences for neonates. Probiotics can be effective in limiting the growth of bacteria associated with dysbiosis and promoting the healthy development of the infant microbiome. Given its adaptation to the infant gut, and promising data from animal and in vitro models, Bifidobacterium longum subsp. infantis is an attractive candidate for use in infant probiotics. However, strain-level differences in the ability of commercialized strains to utilize human milk oligosaccharides (HMOs) may have implications in the performance of strains in the infant gut. In this study, we characterized twelve B. infantis probiotic strains and identified two main variants in one of the HMO utilization gene clusters. Some strains possessed the full repertoire of HMO utilization genes (H5-positive strains), while H5-negative strains lack an ABC-type transporter known to bind core HMO structures. H5-positive strains achieved significantly superior growth on lacto-N-tetraose and lacto-N-neotetraose. In vitro, H5-positive strains had a significant fitness advantage over H5-negative strains, which was also observed in vivo in breastfed infants. This work provides evidence of the functional implications of genetic differences among B. infantis strains and highlights that genotype and HMO utilization phenotype should be considered when selecting a strain for probiotic use in infants.


Assuntos
Bifidobacterium longum subspecies infantis/genética , Microbioma Gastrointestinal/genética , Leite Humano/microbiologia , Oligossacarídeos/genética , Probióticos/química , Hibridização Genômica Comparativa , Disbiose/microbiologia , Disbiose/prevenção & controle , Genótipo , Humanos , Recém-Nascido
11.
Int J Mol Sci ; 21(13)2020 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-32610704

RESUMO

Evidence that whey proteins and peptides have health benefits beyond basic infant nutrition has increased dramatically in recent years. Previously, we demonstrated that a whey-derived immunoglobulin G-enriched powder (IGEP) enhanced adhesion of Bifidobacterium longum subsp. infantis ATCC 15697 (B. infantis) to HT-29 cells. In this study, we investigated the synergistic effect of IGEP-treated B. infantis on preventing the attachment of highly invasive Campylobacter jejuni 81-176 (C. jejuni) to intestinal HT-29 cells. The combination decreased the adherence of C. jejuni to the HT-29 cells by an average of 48% compared to the control (non-IGEP-treated B. infantis). We also confirmed that treatment of IGEP with sodium metaperiodate, which disables the biological recognition of the conjugated oligosaccharides, reduced adhesion of B. infantis to the intestinal cells. Thus, glycosylation of the IGEP components may be important in enhancing B. infantis adhesion. Interestingly, an increased adhesion phenotype was not observed when B. infantis was treated with bovine serum-derived IgG, suggesting that bioactivity was unique to milk-derived immunoglobulin-rich powders. Notably, IGEP did not induce growth of B. infantis within a 24 hours incubation period, as demonstrated by growth curves and metabolite analysis. The current study provides insight into the functionality of bovine whey components and highlights their potential in positively impacting the development of a healthy microbiota.


Assuntos
Bifidobacterium longum subspecies infantis/efeitos dos fármacos , Campylobacter jejuni/efeitos dos fármacos , Proteínas do Soro do Leite/farmacologia , Soro do Leite/química , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/metabolismo , Campylobacter jejuni/genética , DNA Bacteriano/genética , Células HT29 , Humanos , Imunoglobulina G/metabolismo , Intestinos/microbiologia , Microbiota/genética , Soro do Leite/metabolismo , Proteínas do Soro do Leite/metabolismo
12.
Sci Rep ; 9(1): 7983, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138818

RESUMO

Human milk oligosaccharides (HMOs) function as prebiotics for beneficial bacteria in the developing gut, often dominated by Bifidobacterium spp. To understand the relationship between bifidobacteria utilizing HMOs and how the metabolites that are produced could affect the host, we analyzed the metabolism of HMO 2'-fucosyllactose (2'-FL) in Bifidobacterium longum subsp. infantis Bi-26. RNA-seq and metabolite analysis (NMR/GCMS) was performed on samples at early (A600 = 0.25), mid-log (0.5-0.7) and late-log phases (1.0-2.0) of growth. Transcriptomic analysis revealed many gene clusters including three novel ABC-type sugar transport clusters to be upregulated in Bi-26 involved in processing of 2'-FL along with metabolism of its monomers glucose, fucose and galactose. Metabolite data confirmed the production of formate, acetate, 1,2-propanediol, lactate and cleaving of fucose from 2'-FL. The formation of acetate, formate, and lactate showed how the cell uses metabolites during fermentation to produce higher levels of ATP (mid-log compared to other stages) or generate cofactors to balance redox. We concluded that 2'-FL metabolism is a complex process involving multiple gene clusters, that produce a more diverse metabolite profile compared to lactose. These results provide valuable insight on the mode-of-action of 2'-FL utilization by Bifidobacterium longum subsp. infantis Bi-26.


Assuntos
Proteínas de Bactérias/genética , Bifidobacterium longum subspecies infantis/metabolismo , Microbioma Gastrointestinal/fisiologia , Leite Humano/química , Transcriptoma , Trissacarídeos/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/biossíntese , Proteínas de Bactérias/metabolismo , Bifidobacterium longum subspecies infantis/genética , Feminino , Fermentação , Fucose/metabolismo , Galactose/metabolismo , Galactosidases/genética , Galactosidases/metabolismo , Glucose/metabolismo , Humanos , Família Multigênica , Prebióticos/análise , Análise de Componente Principal , Simbiose/fisiologia , alfa-L-Fucosidase/genética , alfa-L-Fucosidase/metabolismo
13.
Benef Microbes ; 9(6): 927-935, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30099889

RESUMO

The ban on the use of antibiotics as feed additives for animal growth promotion in the European Union and United States and the expectation of this trend to further expand to other countries in the short term have prompted a surge in probiotic research. Multi-species probiotics including safe and compatible strains with the ability to bind different nutritional lectins with detrimental effects on poultry nutrition could replace antibiotics as feed additives. Lactobacillus salivarius LET201, Lactobacillus reuteri LET210, Enterococcus faecium LET301, Propionibacterium acidipropionici LET103 and Bifidobacterium infantis CRL1395 have proved to be compatible as evaluated through three different approaches: the production and excretion of antimicrobial compounds, growth inhibition by competition for essential nutrients and physical contact, and a combination of both. The safety of P. acidipropionici LET103 was confirmed, since no expression of virulence factors or antibiotic resistance was detected. The innocuity of E. faecium LET301 should be further evaluated, since the presence of genes coding for certain virulence factors (gelE, efaAfm and efaAfs) was observed, albeit no expression of gelE was previously detected for this strain and there are no reports of involvement of efaAfm in animal pathogenicity. Finally, a combination of the five strains effectively protected intestinal epithelial cells of broilers from the cytotoxicity of mixtures of soybean agglutinin, wheat germ agglutinin and concanavalin A. To our knowledge, this is the first time that a combination of strains is evaluated for their protection against lectins that might be simultaneously present in poultry feeds.


Assuntos
Anti-Infecciosos/metabolismo , Bifidobacterium longum subspecies infantis/metabolismo , Enterococcus faecium/metabolismo , Lactobacillus/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Probióticos/farmacologia , Propionibacterium/metabolismo , Animais , Antibiose , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/crescimento & desenvolvimento , Bifidobacterium longum subspecies infantis/patogenicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Concanavalina A/toxicidade , Farmacorresistência Bacteriana , Enterococcus faecium/genética , Enterococcus faecium/crescimento & desenvolvimento , Enterococcus faecium/patogenicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Lactobacillus/genética , Lactobacillus/crescimento & desenvolvimento , Lactobacillus/patogenicidade , Lectinas/metabolismo , Modelos Teóricos , Lectinas de Plantas/toxicidade , Probióticos/efeitos adversos , Propionibacterium/genética , Propionibacterium/crescimento & desenvolvimento , Propionibacterium/patogenicidade , Ligação Proteica , Proteínas de Soja/toxicidade , Virulência , Fatores de Virulência/genética , Aglutininas do Germe de Trigo/toxicidade
14.
Sci Rep ; 8(1): 10750, 2018 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-30013208

RESUMO

Bifidobacteria are beneficial anaerobes, and their O2 sensitivity levels differ among species as a function of unknown molecular mechanisms. Bifidobacterium longum subspecies infantis (B. infantis), a predominant colonizer of the gastrointestinal tract of infants, showed a hyper O2-sensitive growth profile with accompanying a production of H2O2. In this study, we characterized an NADPH oxidase as a key enzyme responsible for this microbe's hyper O2 sensitivity. A dominant active elution peak of H2O2-forming NADPH oxidase activity was detected in the first step of column chromatography, and the purified NADPH oxidase (NPOX) was identified as a homolog of nitroreductase family proteins. The introduction of the gene encoding B. infantis NPOX (npoxA) into O2-tolerant Bifidobacterium minimum made the strain O2 sensitive and allowed it to produce H2O2. Knockout of the npoxA gene in B. infantis decreased the production of H2O2 and mitigated its B. infantis hyper O2 sensitivity. A transcript of B. infantis npoxA is induced by O2, suggesting that the aerobic production of toxic H2O2 is functionally conserved in B. infantis.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium longum subspecies infantis/enzimologia , Peróxido de Hidrogênio/metabolismo , NADPH Oxidases/metabolismo , Oxigênio/toxicidade , Bactérias Anaeróbias/genética , Bactérias Anaeróbias/metabolismo , Proteínas de Bactérias/genética , Bifidobacterium longum subspecies infantis/genética , DNA Bacteriano/genética , Técnicas de Inativação de Genes , NADPH Oxidases/genética , Estresse Oxidativo
15.
J Dairy Sci ; 101(8): 6730-6741, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29803426

RESUMO

Bovine milk glycomacropeptide (GMP) is derived from κ-casein, with exclusively o-linked glycosylation. Glycomacropeptide promoted the growth of Bifidobacterium longum ssp. infantis in a concentration-dependent manner, and this activity was lost following periodate treatment of the GMP (GMP-P), which disables biological recognition of the conjugated oligosaccharides. Transcriptional analysis of B. longum ssp. infantis following exposure to GMP revealed a substantial response to GMP relative to bacteria treated with GMP-P, with a greater number of differentially expressed transcripts and larger fold changes versus the control. Therefore, stimulation of B. longum ssp. infantis growth by GMP is intrinsically linked to the peptide's O-linked glycosylation. The pool of differentially expressed transcripts included 2 glycoside hydrolase (family 25) genes, which were substantially upregulated following exposure to GMP, but not GMP-P. These GH25 genes were present in duplicated genomic islands that also contained genes encoding fibronectin type III binding domain proteins and numerous phage-related proteins, all of which were also upregulated. Homologs of this genomic arrangement were present in other Bifidobacterium species, which suggest it may be a conserved domain for the utilization of glycosylated peptides. This study provides insights into the molecular basis for the prebiotic effect of bovine milk GMP on B. longum ssp. infantis.


Assuntos
Bifidobacterium longum subspecies infantis/crescimento & desenvolvimento , Bifidobacterium longum subspecies infantis/genética , Caseínas/farmacologia , Regulação Bacteriana da Expressão Gênica , Fragmentos de Peptídeos/farmacologia , Animais , Bovinos , Oligossacarídeos
17.
J Appl Microbiol ; 121(1): 263-76, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27086652

RESUMO

AIM: In this study, we describe the isolation of a gene encoding a novel ß-fructofuranosidase from Bifidobacterium longum subsp. infantis ATCC 15697, and the characterization of the enzyme, the second one found in this strain, significantly different in primary sequence to the already reported bifidobacterial ß-fructofuranosidases. METHODS AND RESULTS: The gene, found through genome-mining was expressed in Escherichia coli C41(DE3). The recombinant enzyme (B.longum_l1) has a molecular weight of 75 kDa, with optimal activity at 50°C, pH 6·0-6·5, and a remarkable stability with a half-life of 75·5 h at 50°C. B.longum_l1 has a wide specificity for fructans, hydrolysing all substrates through an exo-type mechanism, including Oligofructose P95 (ß2-1 fructooligosaccharides (FOS), DP 2-8), Raftilose Synergy 1(ß2-1 FOS & inulin, DP 2-60), Raftiline HP (inulin, DP 2-60), bacterial inulin (3000 kDa) and levan (8·3 & 3500 kDa), Agave fructans (mixed fructans, DP 3-29) and levan-type FOS (ß2-6 FOS, DP 2-8), with the highest relative activity and turnover number found for levan-type FOS. The apparent affinity of the enzyme for levan-type FOS and Oligofructose P95 was found to be 9·2 and 4·6 mmol l(-1) (Km ) with a specific activity of 908 and 725 µmol min(-1)  mg(-1) of protein (k2 ), respectively, more than twice the activity for sucrose. CONCLUSION: B.longum_l1 is a wide substrate specificity enzyme, which may contribute to the competitiveness and persistence of this strain in the colon. SIGNIFICANCE AND IMPACT OF THE STUDY: The bifidobacterial ß-fructofuranosidase activity was evaluated with a wide variety of substrates including noncommercial fructans, such as levan-type and mixed agave fructans. Its activity on these substrates certainly strengthens their commercial prebiotic character and contributes to the understanding of bifidobacteria stimulation by structurally diverse fructans.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium longum subspecies infantis/enzimologia , Frutanos/química , Frutanos/metabolismo , beta-Frutofuranosidase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/metabolismo , Peso Molecular , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Especificidade por Substrato , Sacarose/metabolismo , beta-Frutofuranosidase/química , beta-Frutofuranosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...